1
|
Woodland JG, Coertzen D, Wicht KJ, Hidalgo VF, Pasaje CFA, Godoy LC, Qahash T, Mmonwa MM, Dziwornu GA, Wambua L, Harries S, Korkor CM, Njoroge M, Krugmann L, Taylor D, Leshabane M, Langeveld H, Rabie T, Reader J, van der Watt M, Venter N, Erlank E, Aswat AS, Koekemoer LL, Yeo T, Jeon JH, Fidock DA, Gamo FJ, Wittlin S, Niles JC, Llinas M, Coulson LB, Birkholtz LM, Chibale K. The ATM Kinase Inhibitor AZD0156 Is a Potent Inhibitor of Plasmodium Phosphatidylinositol 4-Kinase (PI4Kβ) and Is an Attractive Candidate for Medicinal Chemistry Optimization Against Malaria. Angew Chem Int Ed Engl 2025:e202425206. [PMID: 40317875 DOI: 10.1002/anie.202425206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/13/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
New compounds targeting human malaria parasites are critical for effective malaria control and elimination. Here, we pursued the imidazoquinolinone AZD0156 (MMV1580483), a human ataxia-telangiectasia mutated (ATM) kinase inhibitor that completed Phase I clinical trials as an anticancer agent. We validated its in vitro activity against the two main forms of the Plasmodium falciparum parasite in the human host, viz. the asexual blood (symptomatic) stage and sexual gametocyte (transmission) stage. Resistance selection, cross-resistance, biochemical, and conditional knockdown studies revealed that AZD0156 inhibits P. falciparum phosphatidylinositol 4-kinase type III beta (PfPI4Kβ), a clinically-validated target for the treatment of malaria. Metabolic perturbations, fixed-ratio isobolograms, killing kinetics and morphological evaluation correlated AZD0156 inhibition with other known PI4Kβ inhibitors. The compound showed favorable in vivo pharmacokinetic properties and 81% antimalarial efficacy (4 × 50 mg kg-1) in a P. berghei mouse malaria infection model. Importantly, a cleaner biochemical profile was measured against human kinases (MAP4K4, MINK1) implicated in embryofoetal developmental toxicity associated with the PfPI4Kβ inhibitor MMV390048. This improved kinase selectivity profile and structural differentiation from other PI4Kβ inhibitors, together with its multistage antiplasmodial activity and favorable pharmacokinetic properties, makes AZD0156 an attractive candidate for target-based drug repositioning against malaria via a medicinal chemistry optimization approach.
Collapse
Affiliation(s)
- John G Woodland
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, 7925, South Africa
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
| | - Dina Coertzen
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, 0028, South Africa
| | - Kathryn J Wicht
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, 7925, South Africa
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
| | | | - Charisse Flerida A Pasaje
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Luiz C Godoy
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Tarrick Qahash
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, State College, PA, 16802, USA
- Huck Center for Malaria Research, Pennsylvania State University, University Park, State College, PA, 16802, USA
| | - Mmakwena M Mmonwa
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
| | - Godwin A Dziwornu
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
| | - Lynn Wambua
- South African Medical Research Council Drug Discovery and Development Research Unit, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, 7925, South Africa
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
| | - Sarah Harries
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
| | - Constance M Korkor
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
| | - Mathew Njoroge
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
| | - Liezl Krugmann
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
| | - Dale Taylor
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
| | - Meta Leshabane
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, 0028, South Africa
| | - Henrico Langeveld
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, 0028, South Africa
| | - Tayla Rabie
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, 0028, South Africa
| | - Janette Reader
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, 0028, South Africa
| | - Mariëtte van der Watt
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, 0028, South Africa
| | - Nelius Venter
- Wits Research Institute for Malaria, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
- Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases, a Division of the National Health Laboratory Service, Johannesburg, 2193, South Africa
| | - Erica Erlank
- Wits Research Institute for Malaria, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
- Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases, a Division of the National Health Laboratory Service, Johannesburg, 2193, South Africa
| | - Ayesha S Aswat
- Wits Research Institute for Malaria, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
- Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases, a Division of the National Health Laboratory Service, Johannesburg, 2193, South Africa
| | - Lizette L Koekemoer
- Wits Research Institute for Malaria, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
- Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases, a Division of the National Health Laboratory Service, Johannesburg, 2193, South Africa
| | - Tomas Yeo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jin H Jeon
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | | | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, Allschwil, 4123, Switzerland
- University of Basel, Basel, 4003, Switzerland
| | - Jacquin C Niles
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Manuel Llinas
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, State College, PA, 16802, USA
- Huck Center for Malaria Research, Pennsylvania State University, University Park, State College, PA, 16802, USA
| | - Lauren B Coulson
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, 7925, South Africa
| | - Lyn-Marié Birkholtz
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, 0028, South Africa
- Department of Biochemistry, Stellenbosch University, Stellenbosch, Matieland, 7602, South Africa
| | - Kelly Chibale
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, 7925, South Africa
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
| |
Collapse
|
2
|
Jia X, Wang Y, Wang M, Min H, Fang Z, Lu H, Li J, Cao Y, Bai L, Lu J. The phosphatase inhibitor BVT-948 can be used to efficiently screen functional sexual development proteins in the malaria parasite Plasmodium berghei. Int J Parasitol Drugs Drug Resist 2024; 26:100563. [PMID: 39153438 PMCID: PMC11378252 DOI: 10.1016/j.ijpddr.2024.100563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Studying and discovering the molecular mechanism of Plasmodium sexual development is crucial for the development of transmission blocking drugs and malaria eradication. The aim of this study was to investigate the feasibility of using phosphatase inhibitors as a tool for screening proteins essential for Plasmodium sexual development and to discover proteins affecting the sexual development of malaria parasites. METHODS Differences in protein phosphorylation among Plasmodium gametocytes incubated with BVT-948 under in vitro ookinete culture conditions were evaluated using phosphoproteomic methods. Gene Ontology (GO) analysis was performed to predict the mechanism by which BVT-948 affected gametocyte-ookinete conversion. The functions of 8 putative proteins involved in Plasmodium berghei sexual development were evaluated. Bioinformatic analysis was used to evaluate the possible mechanism of PBANKA_0100800 in gametogenesis and subsequent sexual development. RESULTS The phosphorylation levels of 265 proteins decreased while those of 67 increased after treatment with BVT-948. Seven of the 8 genes selected for phenotype screening play roles in P. berghei sexual development, and 4 of these were associated with gametocytogenesis. PBANKA_0100800 plays essential roles in gametocyte-ookinete conversion and transmission to mosquitoes. CONCLUSIONS Seven proteins identified by screening affect P. berghei sexual development, suggesting that phosphatase inhibitors can be used for functional protein screening.
Collapse
Affiliation(s)
- Xitong Jia
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, 110000, China; Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110122, China
| | - Yong Wang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110122, China; Department of Family Medicine, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Meilian Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110122, China
| | - Hui Min
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110122, China
| | - Zehou Fang
- The Second Clinical College of China Medical University, Shenyang, Liaoning, 110122, China
| | - Haifeng Lu
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110122, China
| | - Jiao Li
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110122, China
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110122, China.
| | - Lunhao Bai
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, 110000, China.
| | - Jinghan Lu
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, 110000, China.
| |
Collapse
|
3
|
Naude M, van Heerden A, Reader J, van der Watt M, Niemand J, Joubert D, Siciliano G, Alano P, Njoroge M, Chibale K, Herreros E, Leroy D, Birkholtz LM. Eliminating malaria transmission requires targeting immature and mature gametocytes through lipoidal uptake of antimalarials. Nat Commun 2024; 15:9896. [PMID: 39548094 PMCID: PMC11568134 DOI: 10.1038/s41467-024-54144-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024] Open
Abstract
Novel antimalarial compounds targeting both the pathogenic and transmissible stages of the human malaria parasite, Plasmodium falciparum, would greatly benefit malaria elimination strategies. However, most compounds affecting asexual blood stage parasites show severely reduced activity against gametocytes. The impact of this activity loss on a compound's transmission-blocking activity is unclear. Here, we report the systematic evaluation of the activity loss against gametocytes and investigate the confounding factors contributing to this. A threshold for acceptable activity loss between asexual blood stage parasites and gametocytes was defined, with near-equipotent compounds required to prevent continued gametocyte maturation and onward transmission. Target abundance is not predictive of gametocytocidal activity, but instead, lipoidal uptake is the main barrier of dual activity and is influenced by distinct physicochemical properties. This study provides guidelines for the required profiles of potential dual-active antimalarial agents and facilitates the development of effective transmission-blocking compounds.
Collapse
Affiliation(s)
- Mariska Naude
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
- Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
| | - Ashleigh van Heerden
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
- Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
| | - Janette Reader
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
- Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
| | - Mariëtte van der Watt
- Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
| | - Jandeli Niemand
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
- Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
| | - Dorè Joubert
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
| | - Giulia Siciliano
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| | - Pietro Alano
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| | - Mathew Njoroge
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
| | - Kelly Chibale
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
| | | | - Didier Leroy
- Medicines for Malaria Venture, Geneva, Switzerland
| | - Lyn-Marié Birkholtz
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria, 0028, South Africa.
- Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria, 0028, South Africa.
- Department of Biochemistry, Stellenbosch University, Matieland, Stellenbosch, 7602, South Africa.
| |
Collapse
|
4
|
Watson SJ, van der Watt ME, Theron A, Reader J, Tshabalala S, Erlank E, Koekemoer LL, Naude M, Stampolaki M, Adewole F, Sadowska K, Pérez-Lozano P, Turcu AL, Vázquez S, Ko J, Mazurek B, Singh D, Malwal SR, Njoroge M, Chibale K, Onajole OK, Kolocouris A, Oldfield E, Birkholtz LM. The Tuberculosis Drug Candidate SQ109 and Its Analogs Have Multistage Activity against Plasmodium falciparum. ACS Infect Dis 2024; 10:3358-3367. [PMID: 39143042 PMCID: PMC11406516 DOI: 10.1021/acsinfecdis.4c00461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Toward repositioning the antitubercular clinical candidate SQ109 as an antimalarial, analogs were investigated for structure-activity relationships for activity against asexual blood stages of the human malaria parasite Plasmodium falciparum pathogenic forms, as well as transmissible, sexual stage gametocytes. We show that equipotent activity (IC50) in the 100-300 nM range could be attained for both asexual and sexual stages, with the activity of most compounds retained against a multidrug-resistant strain. The multistage activity profile relies on high lipophilicity ascribed to the adamantane headgroup, and antiplasmodial activity is critically dependent on the diamine linker. Frontrunner compounds showed conserved activity against genetically diverse southern African clinical isolates. We additionally validated that this series could block transmission to mosquitoes, marking these compounds as novel chemotypes with multistage antiplasmodial activity.
Collapse
Affiliation(s)
| | | | - Anjo Theron
- Next Generation Health, Council for Scientific and Industrial Research, Pretoria 0001, South Africa
| | | | | | - Erica Erlank
- Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Services, Wits Research Institute for Malaria, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Johannesburg 2000, South Africa
| | - Lizette L Koekemoer
- Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Services, Wits Research Institute for Malaria, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Johannesburg 2000, South Africa
| | | | - Marianna Stampolaki
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis-Zografou, 15771 Athens, Greece
| | - Feyisola Adewole
- Department of Biological, Physical and Health Sciences, College of Science, Health & Pharmacy, Roosevelt University, 425 South Wabash Avenue, Chicago, Illinois 60605, United States
| | - Katie Sadowska
- Department of Biological, Physical and Health Sciences, College of Science, Health & Pharmacy, Roosevelt University, 425 South Wabash Avenue, Chicago, Illinois 60605, United States
| | - Pilar Pérez-Lozano
- Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona 08028, Spain
| | - Andreea L Turcu
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, and Institute of Biomedicine (IBUB), Universitat de Barcelona, Barcelona E-08028, Spain
| | - Santiago Vázquez
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, and Institute of Biomedicine (IBUB), Universitat de Barcelona, Barcelona E-08028, Spain
| | - Jihee Ko
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Ben Mazurek
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Davinder Singh
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Satish R Malwal
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Mathew Njoroge
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch, Capetown 7701, South Africa
| | - Kelly Chibale
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch, Capetown 7701, South Africa
- South African Medical Research Council Drug Discovery and Development Centre, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, Capetown 7701, South Africa
| | - Oluseye K Onajole
- Department of Biological, Physical and Health Sciences, College of Science, Health & Pharmacy, Roosevelt University, 425 South Wabash Avenue, Chicago, Illinois 60605, United States
| | - Antonios Kolocouris
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis-Zografou, 15771 Athens, Greece
| | - Eric Oldfield
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | | |
Collapse
|
5
|
Lokole PB, Byamungu GG, Mutwale PK, Ngombe NK, Mudogo CN, Krause RWM, Nkanga CI. Plant-based nanoparticles targeting malaria management. Front Pharmacol 2024; 15:1440116. [PMID: 39185312 PMCID: PMC11341498 DOI: 10.3389/fphar.2024.1440116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/18/2024] [Indexed: 08/27/2024] Open
Abstract
Malaria is one of the most devastating diseases across the globe, particularly in low-income countries in Sub-Saharan Africa. The increasing incidence of malaria morbidity is mainly due to the shortcomings of preventative measures such as the lack of vaccines and inappropriate control over the parasite vector. Additionally, high mortality rates arise from therapeutic failures due to poor patient adherence and drug resistance development. Although the causative pathogen (Plasmodium spp.) is an intracellular parasite, the recommended antimalarial drugs show large volumes of distribution and low-to no-specificity towards the host cell. This leads to severe side effects that hamper patient compliance and promote the emergence of drug-resistant strains. Recent research efforts are promising to enable the discovery of new antimalarial agents; however, the lack of efficient means to achieve targeted delivery remains a concern, given the risk of further resistance development. New strategies based on green nanotechnologies are a promising avenue for malaria management due to their potential to eliminate malaria vectors (Anopheles sp.) and to encapsulate existing and emerging antimalarial agents and deliver them to different target sites. In this review we summarized studies on the use of plant-derived nanoparticles as cost-effective preventative measures against malaria parasites, starting from the vector stage. We also reviewed plant-based nanoengineering strategies to target malaria parasites, and further discussed the site-specific delivery of natural products using ligand-decorated nanoparticles that act through receptors on the host cells or malaria parasites. The exploration of traditionally established plant medicines, surface-engineered nanoparticles and the molecular targets of parasite/host cells may provide valuable insights for future discovery of antimalarial drugs and open new avenues for advancing science toward the goal of malaria eradication.
Collapse
Affiliation(s)
- Pathy B. Lokole
- Centre de Recherche en Nanotechnologies Appliquées aux Produits Naturels (CReNAPN), Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
- Centre d’Etudes des Substances Naturelles d’Origine Végétale (CESNOV), Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
- Center for Chemico- and Bio-Medicinal Research (CCBR), Department of Chemistry, Faculty of Sciences, Rhodes University, Grahamstown, Eastern Cape, South Africa
| | - Galilée G. Byamungu
- Centre de Recherche en Nanotechnologies Appliquées aux Produits Naturels (CReNAPN), Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
- Center for Chemico- and Bio-Medicinal Research (CCBR), Department of Chemistry, Faculty of Sciences, Rhodes University, Grahamstown, Eastern Cape, South Africa
- Department of Chemistry, Faculty of Sciences and Technology, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Paulin K. Mutwale
- Centre de Recherche en Nanotechnologies Appliquées aux Produits Naturels (CReNAPN), Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
- Centre d’Etudes des Substances Naturelles d’Origine Végétale (CESNOV), Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Nadège K. Ngombe
- Centre de Recherche en Nanotechnologies Appliquées aux Produits Naturels (CReNAPN), Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
- Centre d’Etudes des Substances Naturelles d’Origine Végétale (CESNOV), Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Celestin N. Mudogo
- Unit of Molecular Biology, Department of Basic Sciences, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Rui W. M. Krause
- Center for Chemico- and Bio-Medicinal Research (CCBR), Department of Chemistry, Faculty of Sciences, Rhodes University, Grahamstown, Eastern Cape, South Africa
| | - Christian I. Nkanga
- Centre de Recherche en Nanotechnologies Appliquées aux Produits Naturels (CReNAPN), Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| |
Collapse
|
6
|
Appetecchia F, Fabbrizi E, Fiorentino F, Consalvi S, Biava M, Poce G, Rotili D. Transmission-Blocking Strategies for Malaria Eradication: Recent Advances in Small-Molecule Drug Development. Pharmaceuticals (Basel) 2024; 17:962. [PMID: 39065810 PMCID: PMC11279868 DOI: 10.3390/ph17070962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Malaria drug research and development efforts have resurged in the last decade following the decelerating rate of mortality and malaria cases in endemic regions. The inefficiency of malaria interventions is largely driven by the spreading resistance of the Plasmodium falciparum parasite to current drug regimens and that of the malaria vector, the Anopheles mosquito, to insecticides. In response to the new eradication agenda, drugs that act by breaking the malaria transmission cycle (transmission-blocking drugs), which has been recognized as an important and additional target for intervention, are being developed. These drugs take advantage of the susceptibility of Plasmodium during population bottlenecks before transmission (gametocytes) and in the mosquito vector (gametes, zygotes, ookinetes, oocysts, sporozoites). To date, compounds targeting stage V gametocytes predominate in the chemical library of transmission-blocking drugs, and some of them have entered clinical trials. The targeting of Plasmodium mosquito stages has recently renewed interest in the development of innovative malaria control tools, which hold promise for the application of compounds effective at these stages. In this review, we highlight the major achievements and provide an update on the research of transmission-blocking drugs, with a particular focus on their chemical scaffolds, antiplasmodial activity, and transmission-blocking potential.
Collapse
Affiliation(s)
| | | | | | | | | | - Giovanna Poce
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (F.A.); (E.F.); (F.F.); (S.C.); (M.B.)
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (F.A.); (E.F.); (F.F.); (S.C.); (M.B.)
| |
Collapse
|
7
|
Dziwornu G, Seanego D, Fienberg S, Clements M, Ferreira J, Sypu VS, Samanta S, Bhana AD, Korkor CM, Garnie LF, Teixeira N, Wicht KJ, Taylor D, Olckers R, Njoroge M, Gibhard L, Salomane N, Wittlin S, Mahato R, Chakraborty A, Sevilleno N, Coyle R, Lee MCS, Godoy LC, Pasaje CF, Niles JC, Reader J, van der Watt M, Birkholtz LM, Bolscher JM, de Bruijni MHC, Coulson LB, Basarab GS, Ghorpade SR, Chibale K. 2,8-Disubstituted-1,5-naphthyridines as Dual Inhibitors of Plasmodium falciparum Phosphatidylinositol-4-kinase and Hemozoin Formation with In Vivo Efficacy. J Med Chem 2024; 67:11401-11420. [PMID: 38918002 PMCID: PMC11247499 DOI: 10.1021/acs.jmedchem.4c01154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024]
Abstract
Structure-activity relationship studies of 2,8-disubstituted-1,5-naphthyridines, previously reported as potent inhibitors of Plasmodium falciparum (Pf) phosphatidylinositol-4-kinase β (PI4K), identified 1,5-naphthyridines with basic groups at 8-position, which retained Plasmodium PI4K inhibitory activity but switched primary mode of action to the host hemoglobin degradation pathway through inhibition of hemozoin formation. These compounds showed minimal off-target inhibitory activity against the human phosphoinositide kinases and MINK1 and MAP4K kinases, which were associated with the teratogenicity and testicular toxicity observed in rats for the PfPI4K inhibitor clinical candidate MMV390048. A representative compound from the series retained activity against field isolates and lab-raised drug-resistant strains of Pf. It was efficacious in the humanized NSG mouse malaria infection model at a single oral dose of 32 mg/kg. This compound was nonteratogenic in the zebrafish embryo model of teratogenicity and has a low predicted human dose, indicating that this series has the potential to deliver a preclinical candidate for malaria.
Collapse
Affiliation(s)
- Godwin
Akpeko Dziwornu
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Donald Seanego
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Stephen Fienberg
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Monica Clements
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Jasmin Ferreira
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Venkata S. Sypu
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Sauvik Samanta
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Ashlyn D. Bhana
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Constance M. Korkor
- Department
of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Larnelle F. Garnie
- Department
of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Nicole Teixeira
- Department
of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Kathryn J. Wicht
- Drug
Discovery and Development Centre (H3D), Department of Chemistry and
Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Dale Taylor
- Drug
Discovery and Development Centre (H3D), Division of Clinical Pharmacology,
Department of Medicine, University of Cape
Town, Observatory 7925, South Africa
| | - Ronald Olckers
- Drug
Discovery and Development Centre (H3D), Division of Clinical Pharmacology,
Department of Medicine, University of Cape
Town, Observatory 7925, South Africa
| | - Mathew Njoroge
- Drug
Discovery and Development Centre (H3D), Division of Clinical Pharmacology,
Department of Medicine, University of Cape
Town, Observatory 7925, South Africa
| | - Liezl Gibhard
- Drug
Discovery and Development Centre (H3D), Division of Clinical Pharmacology,
Department of Medicine, University of Cape
Town, Observatory 7925, South Africa
| | - Nicolaas Salomane
- Drug
Discovery and Development Centre (H3D), Institute of Infectious Disease
and Molecular Medicine, University of Cape
Town, Observatory, Cape Town 7925, South Africa
| | - Sergio Wittlin
- Swiss Tropical
and Public Health Institute, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- University
of Basel, 4001 Basel, Switzerland
| | | | | | - Nicole Sevilleno
- Wellcome
Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, U.K.
| | - Rachael Coyle
- Wellcome
Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, U.K.
| | - Marcus C. S. Lee
- Wellcome
Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, U.K.
| | - Luiz C. Godoy
- Department
of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Charisse Flerida Pasaje
- Department
of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Jacquin C. Niles
- Department
of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Janette Reader
- Department
of Biochemistry, Genetics and Microbiology, Institute
for Sustainable Malaria Control, University
of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Mariette van der Watt
- Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Lyn-Marié Birkholtz
- Department
of Biochemistry, Genetics and Microbiology, Institute
for Sustainable Malaria Control, University
of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Judith M. Bolscher
- TropIQ Health Sciences, Transistorweg 5, 6534 AT Nijmegen, The Netherlands
| | | | - Lauren B. Coulson
- Drug
Discovery and Development Centre (H3D), Institute of Infectious Disease
and Molecular Medicine, University of Cape
Town, Observatory, Cape Town 7925, South Africa
| | - Gregory S. Basarab
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- Drug
Discovery and Development Centre (H3D), Division of Clinical Pharmacology,
Department of Medicine, University of Cape
Town, Observatory 7925, South Africa
| | - Sandeep R. Ghorpade
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Kelly Chibale
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- South African Medical Research Council Drug Discovery
and Development
Research Unit, Department of Chemistry and Institute of Infectious
Disease and Molecular Medicine, University
of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|
8
|
Vareta J, Horstman NA, Adams M, Seydel KB, McCann RS, Cohee LM, Laufer MK, Takala-Harrison S. Genotyping Plasmodium falciparum gametocytes using amplicon deep sequencing. Malar J 2024; 23:96. [PMID: 38582837 PMCID: PMC10999092 DOI: 10.1186/s12936-024-04920-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 03/27/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Understanding the dynamics of gametocyte production in polyclonal Plasmodium falciparum infections requires a genotyping method that detects distinct gametocyte clones and estimates their relative frequencies. Here, a marker was identified and evaluated to genotype P. falciparum mature gametocytes using amplicon deep sequencing. METHODS A data set of polymorphic regions of the P. falciparum genome was mined to identify a gametocyte genotyping marker. To assess marker resolution, the number of unique haplotypes in the marker region was estimated from 95 Malawian P. falciparum whole genome sequences. Specificity of the marker for detection of mature gametocytes was evaluated using reverse transcription-polymerase chain reaction of RNA extracted from NF54 mature gametocytes and rings from a non-gametocyte-producing strain of P. falciparum. Amplicon deep sequencing was performed on experimental mixtures of mature gametocytes from two distinct parasite clones, as well as gametocyte-positive P. falciparum field isolates to evaluate the quantitative ability and determine the limit of detection of the genotyping approach. RESULTS A 400 bp region of the pfs230 gene was identified as a gametocyte genotyping marker. A larger number of unique haplotypes was observed at the pfs230 marker (34) compared to the sera-2 (18) and ama-1 (14) markers in field isolates from Malawi. RNA and DNA genotyping accurately estimated gametocyte and total parasite clone frequencies when evaluating agreement between expected and observed haplotype frequencies in gametocyte mixtures, with concordance correlation coefficients of 0.97 [95% CI: 0.92-0.99] and 0.92 [95% CI: 0.83-0.97], respectively. The detection limit of the genotyping method for male gametocytes was 0.41 pfmget transcripts/µl [95% CI: 0.28-0.72] and for female gametocytes was 1.98 ccp4 transcripts/µl [95% CI: 1.35-3.68]. CONCLUSIONS A region of the pfs230 gene was identified as a marker to genotype P. falciparum gametocytes. Amplicon deep sequencing of this marker can be used to estimate the number and relative frequency of parasite clones among mature gametocytes within P. falciparum infections. This gametocyte genotyping marker will be an important tool for studies aimed at understanding dynamics of gametocyte production in polyclonal P. falciparum infections.
Collapse
Affiliation(s)
- Jimmy Vareta
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Natalie A Horstman
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew Adams
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Karl B Seydel
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - Robert S McCann
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lauren M Cohee
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Miriam K Laufer
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Shannon Takala-Harrison
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
9
|
Greyling N, van der Watt M, Gwarinda H, van Heerden A, Greenhouse B, Leroy D, Niemand J, Birkholtz LM. Genetic complexity alters drug susceptibility of asexual and gametocyte stages of Plasmodium falciparum to antimalarial candidates. Antimicrob Agents Chemother 2024; 68:e0129123. [PMID: 38259087 PMCID: PMC10916389 DOI: 10.1128/aac.01291-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024] Open
Abstract
Malaria elimination requires interventions able to target both the asexual blood stage (ABS) parasites and transmissible gametocyte stages of Plasmodium falciparum. Lead antimalarial candidates are evaluated against clinical isolates to address key concerns regarding efficacy and to confirm that the current, circulating parasites from endemic regions lack resistance against these candidates. While this has largely been performed on ABS parasites, limited data are available on the transmission-blocking efficacy of compounds with multistage activity. Here, we evaluated the efficacy of lead antimalarial candidates against both ABS parasites and late-stage gametocytes side-by-side, against clinical P. falciparum isolates from southern Africa. We additionally correlated drug efficacy to the genetic diversity of the clinical isolates as determined with a panel of well-characterized, genome-spanning microsatellite markers. Our data indicate varying sensitivities of the isolates to key antimalarial candidates, both for ABS parasites and gametocyte stages. While ABS parasites were efficiently killed, irrespective of genetic complexity, antimalarial candidates lost some gametocytocidal efficacy when the gametocytes originated from genetically complex, multiple-clone infections. This suggests a fitness benefit to multiclone isolates to sustain transmission and reduce drug susceptibility. In conclusion, this is the first study to investigate the efficacy of antimalarial candidates on both ABS parasites and gametocytes from P. falciparum clinical isolates where the influence of parasite genetic complexity is highlighted, ultimately aiding the malaria elimination agenda.
Collapse
Affiliation(s)
- Nicola Greyling
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
- Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| | - Mariëtte van der Watt
- Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| | - Hazel Gwarinda
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
- Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| | - Ashleigh van Heerden
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
- Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| | - Bryan Greenhouse
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Didier Leroy
- Medicines for Malaria Venture, Geneva, Switzerland
| | - Jandeli Niemand
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
- Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| | - Lyn-Marié Birkholtz
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
- Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
10
|
van Heerden A, Turon G, Duran-Frigola M, Pillay N, Birkholtz LM. Machine Learning Approaches Identify Chemical Features for Stage-Specific Antimalarial Compounds. ACS OMEGA 2023; 8:43813-43826. [PMID: 38027377 PMCID: PMC10666252 DOI: 10.1021/acsomega.3c05664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/18/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023]
Abstract
Efficacy data from diverse chemical libraries, screened against the various stages of the malaria parasite Plasmodium falciparum, including asexual blood stage (ABS) parasites and transmissible gametocytes, serve as a valuable reservoir of information on the chemical space of compounds that are either active (or not) against the parasite. We postulated that this data can be mined to define chemical features associated with the sole ABS activity and/or those that provide additional life cycle activity profiles like gametocytocidal activity. Additionally, this information could provide chemical features associated with inactive compounds, which could eliminate any future unnecessary screening of similar chemical analogs. Therefore, we aimed to use machine learning to identify the chemical space associated with stage-specific antimalarial activity. We collected data from various chemical libraries that were screened against the asexual (126 374 compounds) and sexual (gametocyte) stages of the parasite (93 941 compounds), calculated the compounds' molecular fingerprints, and trained machine learning models to recognize stage-specific active and inactive compounds. We were able to build several models that predict compound activity against ABS and dual activity against ABS and gametocytes, with Support Vector Machines (SVM) showing superior abilities with high recall (90 and 66%) and low false-positive predictions (15 and 1%). This allowed the identification of chemical features enriched in active and inactive populations, an important outcome that could be mined for essential chemical features to streamline hit-to-lead optimization strategies of antimalarial candidates. The predictive capabilities of the models held true in diverse chemical spaces, indicating that the ML models are therefore robust and can serve as a prioritization tool to drive and guide phenotypic screening and medicinal chemistry programs.
Collapse
Affiliation(s)
- Ashleigh van Heerden
- Department
of Biochemistry, Genetics and Microbiology, Institute for Sustainable
Malaria Control, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Gemma Turon
- Ersilia
Open Source Initiative, 28 Belgrave Road, Cambridge CB1 3DE, U.K.
| | | | - Nelishia Pillay
- Department
of Computer Science, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Lyn-Marié Birkholtz
- Department
of Biochemistry, Genetics and Microbiology, Institute for Sustainable
Malaria Control, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| |
Collapse
|
11
|
Bekić V, Kilian N. Novel secretory organelles of parasite origin - at the center of host-parasite interaction. Bioessays 2023; 45:e2200241. [PMID: 37518819 DOI: 10.1002/bies.202200241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 08/01/2023]
Abstract
Reorganization of cell organelle-deprived host red blood cells by the apicomplexan malaria parasite Plasmodium falciparum enables their cytoadherence to endothelial cells that line the microvasculature. This increases the time red blood cells infected with mature developmental stages remain within selected organs such as the brain to avoid the spleen passage, which can lead to severe complications and cumulate in patient death. The Maurer's clefts are a novel secretory organelle of parasite origin established by the parasite in the cytoplasm of the host red blood cell in order to facilitate the establishment of cytoadherence by conducting the trafficking of immunovariant adhesins to the host cell surface. Another important function of the organelle is the sorting of other proteins the parasite traffics into its host cell. Although the organelle is of high importance for the pathology of malaria, additional putative functions, structure, and genesis remain shrouded in mystery more than a century after its discovery. In this review, we highlight our current knowledge about the Maurer's clefts and other novel secretory organelles established within the host cell cytoplasm by human-pathogenic malaria parasites and other parasites that reside within human red blood cells.
Collapse
Affiliation(s)
- Viktor Bekić
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Nicole Kilian
- Centre for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| |
Collapse
|
12
|
Miyazaki Y, Vos MW, Geurten FJA, Bigeard P, Kroeze H, Yoshioka S, Arisawa M, Inaoka DK, Soulard V, Dechering KJ, Franke-Fayard B, Miyazaki S. A versatile Plasmodium falciparum reporter line expressing NanoLuc enables highly sensitive multi-stage drug assays. Commun Biol 2023; 6:713. [PMID: 37438491 DOI: 10.1038/s42003-023-05078-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 06/27/2023] [Indexed: 07/14/2023] Open
Abstract
Transgenic luciferase-expressing Plasmodium falciparum parasites have been widely used for the evaluation of anti-malarial compounds. Here, to screen for anti-malarial drugs effective against multiple stages of the parasite, we generate a P. falciparum reporter parasite that constitutively expresses NanoLuciferase (NanoLuc) throughout its whole life cycle. The NanoLuc-expressing P. falciparum reporter parasite shows a quantitative NanoLuc signal in the asexual blood, gametocyte, mosquito, and liver stages. We also establish assay systems to evaluate the anti-malarial activity of compounds at the asexual blood, gametocyte, and liver stages, and then determine the 50% inhibitory concentration (IC50) value of several anti-malarial compounds. Through the development of this robust high-throughput screening system, we identify an anti-malarial compound that kills the asexual blood stage parasites. Our study highlights the utility of the NanoLuc reporter line, which may advance anti-malarial drug development through the improved screening of compounds targeting the human malarial parasite at multiple stages.
Collapse
Affiliation(s)
- Yukiko Miyazaki
- Department of Molecular Infection Dynamics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 852-8523, Nagasaki, Japan.
- Department of Parasitology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands.
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.
| | - Martijn W Vos
- TropIQ Health Sciences, Transistorweg 5, 6534 AT, Nijmegen, The Netherlands
| | - Fiona J A Geurten
- Department of Parasitology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Pierre Bigeard
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, F-75013, Paris, France
| | - Hans Kroeze
- Department of Parasitology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Shohei Yoshioka
- Graduate School of Pharmaceutical Sciences, Osaka University, 565-0871, Osaka, Japan
| | - Mitsuhiro Arisawa
- Graduate School of Pharmaceutical Sciences, Osaka University, 565-0871, Osaka, Japan
| | - Daniel Ken Inaoka
- Department of Molecular Infection Dynamics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 852-8523, Nagasaki, Japan
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, 852-8523, Japan
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Valerie Soulard
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, F-75013, Paris, France
| | - Koen J Dechering
- TropIQ Health Sciences, Transistorweg 5, 6534 AT, Nijmegen, The Netherlands
| | - Blandine Franke-Fayard
- Department of Parasitology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Shinya Miyazaki
- Department of Parasitology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands.
- Department of Cellular Architecture Studies, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 852-8523, Nagasaki, Japan.
| |
Collapse
|
13
|
Reader J, Opperman DFL, van der Watt ME, Theron A, Leshabane M, da Rocha S, Turner J, Garrabrant K, Piña I, Mills C, Woster PM, Birkholtz L. New Transmission-Selective Antimalarial Agents through Hit-to-Lead Optimization of 2-([1,1'-Biphenyl]-4-carboxamido)benzoic Acid Derivatives. Chembiochem 2022; 23:e202200427. [PMID: 36106425 PMCID: PMC10946866 DOI: 10.1002/cbic.202200427] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/14/2022] [Indexed: 11/12/2022]
Abstract
Malaria elimination requires multipronged approaches, including the application of antimalarial drugs able to block human-to-mosquito transmission of malaria parasites. The transmissible gametocytes of Plasmodium falciparum seem to be highly sensitive towards epidrugs, particularly those targeting demethylation of histone post-translational marks. Here, we report exploration of compounds from a chemical library generated during hit-to-lead optimization of inhibitors of the human histone lysine demethylase, KDM4B. Derivatives of 2-([1,1'-biphenyl]-4-carboxamido) benzoic acid, around either the amide or a sulfonamide linker backbone (2-(arylcarboxamido)benzoic acid, 2-carboxamide (arylsulfonamido)benzoic acid and N-(2-(1H-tetrazol-5-yl)phenyl)-arylcarboxamide), showed potent activity towards late-stage gametocytes (stage IV/V) of P. falciparum, with the most potent compound reaching single digit nanomolar activity. Structure-activity relationship trends were evident and frontrunner compounds also displayed microsomal stability and favourable solubility profiles. Simplified synthetic routes support further derivatization of these compounds for further development of these series as malaria transmission-blocking agents.
Collapse
Affiliation(s)
- Janette Reader
- Department of BiochemistryGenetics and MicrobiologyInstitute for Sustainable Malaria ControlUniversity of PretoriaLynnwood RoadPretoria0028South Africa
| | - Daniel F. L. Opperman
- Department of BiochemistryGenetics and MicrobiologyInstitute for Sustainable Malaria ControlUniversity of PretoriaLynnwood RoadPretoria0028South Africa
| | - Mariëtte E. van der Watt
- Department of BiochemistryGenetics and MicrobiologyInstitute for Sustainable Malaria ControlUniversity of PretoriaLynnwood RoadPretoria0028South Africa
- School of Health Systems and Public HealthUniversity of Pretoria, HatfieldPretoria0028South Africa
| | - Anjo Theron
- Next Generation HealthCouncil for Scientific and Industrial ResearchPretoria0001South Africa
| | - Meta Leshabane
- Department of BiochemistryGenetics and MicrobiologyInstitute for Sustainable Malaria ControlUniversity of PretoriaLynnwood RoadPretoria0028South Africa
| | - Shanté da Rocha
- Department of BiochemistryGenetics and MicrobiologyInstitute for Sustainable Malaria ControlUniversity of PretoriaLynnwood RoadPretoria0028South Africa
| | - Jonathan Turner
- Department of Drug Discovery and Biomedical SciencesMedical University of South CarolinaCharlestonSC 29425USA
| | - Kathleen Garrabrant
- Department of Drug Discovery and Biomedical SciencesMedical University of South CarolinaCharlestonSC 29425USA
| | - Ivett Piña
- Department of Drug Discovery and Biomedical SciencesMedical University of South CarolinaCharlestonSC 29425USA
| | - Catherine Mills
- Department of Drug Discovery and Biomedical SciencesMedical University of South CarolinaCharlestonSC 29425USA
| | - Patrick M. Woster
- Department of Drug Discovery and Biomedical SciencesMedical University of South CarolinaCharlestonSC 29425USA
| | - Lyn‐Marié Birkholtz
- Department of BiochemistryGenetics and MicrobiologyInstitute for Sustainable Malaria ControlUniversity of PretoriaLynnwood RoadPretoria0028South Africa
| |
Collapse
|
14
|
Reader J, van der Watt ME, Birkholtz LM. Streamlined and Robust Stage-Specific Profiling of Gametocytocidal Compounds Against Plasmodium falciparum. Front Cell Infect Microbiol 2022; 12:926460. [PMID: 35846744 PMCID: PMC9282888 DOI: 10.3389/fcimb.2022.926460] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/30/2022] [Indexed: 12/29/2022] Open
Abstract
Malaria elimination is dependent on the ability to target both the pathogenic and transmissible stages of the human malaria parasite, Plasmodium falciparum. These forms of the parasite are differentiated by unique developmental stages, each with their own biological mechanisms and processes. These individual stages therefore also respond differently to inhibitory compounds, and this complicates the discovery of multistage active antimalarial agents. The search for compounds with transmission-blocking activity has focused on screening for activity on mature gametocytes, with only limited descriptions available for the activity of such compounds on immature stage gametocytes. This therefore poses a gap in the profiling of antimalarial agents for pan-reactive, multistage activity to antimalarial leads. Here, we optimized an effective and robust strategy for the simple and cost-effective description of the stage-specific action of gametocytocidal antimalarial compounds.
Collapse
Affiliation(s)
- Janette Reader
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
| | - Mariette E. van der Watt
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
- Institute for Sustainable Malaria Control, School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa
| | - Lyn-Marié Birkholtz
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
- *Correspondence: Lyn-Marié Birkholtz,
| |
Collapse
|